Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Oncoimmunology ; 12(1): 2184143, 2023.
Article in English | MEDLINE | ID: mdl-36875548

ABSTRACT

Despite massive improvements in the treatment of B-ALL through CART-19 immunotherapy, a large number of patients suffer a relapse due to loss of the targeted epitope. Mutations in the CD19 locus and aberrant splicing events are known to account for the absence of surface antigen. However, early molecular determinants suggesting therapy resistance as well as the time point when first signs of epitope loss appear to be detectable are not enlightened so far. By deep sequencing of the CD19 locus, we identified a blast-specific 2-nucleotide deletion in intron 2 that exists in 35% of B-ALL samples at initial diagnosis. This deletion overlaps with the binding site of RNA binding proteins (RBPs) including PTBP1 and might thereby affect CD19 splicing. Moreover, we could identify a number of other RBPs that are predicted to bind to the CD19 locus being deregulated in leukemic blasts, including NONO. Their expression is highly heterogeneous across B-ALL molecular subtypes as shown by analyzing 706 B-ALL samples accessed via the St. Jude Cloud. Mechanistically, we show that downregulation of PTBP1, but not of NONO, in 697 cells reduces CD19 total protein by increasing intron 2 retention. Isoform analysis in patient samples revealed that blasts, at diagnosis, express increased amounts of CD19 intron 2 retention compared to normal B cells. Our data suggest that loss of RBP functionality by mutations altering their binding motifs or by deregulated expression might harbor the potential for the disease-associated accumulation of therapy-resistant CD19 isoforms.


Subject(s)
Antigens, CD19 , Heterogeneous-Nuclear Ribonucleoproteins , Leukemia, B-Cell , Polypyrimidine Tract-Binding Protein , RNA-Binding Proteins , Humans , Binding Sites , Epitopes , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Mutation , Polypyrimidine Tract-Binding Protein/genetics , RNA-Binding Proteins/genetics , Leukemia, B-Cell/genetics
2.
iScience ; 24(10): 103092, 2021 Oct 22.
Article in English | MEDLINE | ID: mdl-34622147

ABSTRACT

The gut microbiota affects remote organ functions but its impact on organotypic endothelial cell (EC) transcriptomes remains unexplored. The liver endothelium encounters microbiota-derived signals and metabolites via the portal circulation. To pinpoint how gut commensals affect the hepatic sinusoidal endothelium, a magnetic cell sorting protocol, combined with fluorescence-activated cell sorting, was used to isolate hepatic sinusoidal ECs from germ-free (GF) and conventionally raised (CONV-R) mice for transcriptome analysis by RNA sequencing. This resulted in a comprehensive map of microbiota-regulated hepatic EC-specific transcriptome profiles. Gene Ontology analysis revealed that several functional processes in the hepatic endothelium were affected. The absence of microbiota influenced the expression of genes involved in cholesterol flux and angiogenesis. Specifically, genes functioning in hepatic endothelial sphingosine metabolism and the sphingosine-1-phosphate pathway showed drastically increased expression in the GF state. Our analyses reveal a prominent role for the microbiota in shaping the transcriptional landscape of the hepatic endothelium.

3.
Cytometry A ; 99(1): 107-116, 2021 01.
Article in English | MEDLINE | ID: mdl-33090656

ABSTRACT

Results from the first gating proficiency panel of intracellular cytokine staining (ICS) highlighted the value of using a consensus gating approach to reduce the variability across laboratories in reported %CD8+ or %CD4+ cytokine-positive cells. Based on the data analysis from the first proficiency panel, harmonization guidelines for a consensus gating protocol were proposed. To validate the recommendations from the first panel and to examine factors that were not included in the first panel, a second ICS gating proficiency panel was organized. All participants analyzed the same set of Flow Cytometry Standard (FCS) files using their own gating protocol. An optional learning module was provided to demonstrate how to apply the previously established gating recommendations and harmonization guidelines to actual ICS data files. Eighty-three participants took part in this proficiency panel. The results from this proficiency panel confirmed the harmonization guidelines from the first panel. These recommendations addressed the (1) placement of the cytokine-positive gate, (2) identification of CD4+ CD8+ double-positive T cells, (3) placement of lymphocyte gate, (4) inclusion of dim cells, (5) gate uniformity, and (6) proper adjustment of the biexponential scaling. In addition, based on the results of this proficiency gating panel, two new recommendations were added to expand the harmonization guidelines: (1) inclusion of dump channel marker to gate all live and dump negative cells and (2) backgating to confirm the correct placement of gates across all populations. © 2020 International Society for Advancement of Cytometry.


Subject(s)
Cytokines , Neoplasms , Flow Cytometry , Humans , Immunotherapy , Neoplasms/therapy , Reproducibility of Results , Staining and Labeling
4.
Nature ; 585(7823): 107-112, 2020 09.
Article in English | MEDLINE | ID: mdl-32728218

ABSTRACT

Treating patients who have cancer with vaccines that stimulate a targeted immune response is conceptually appealing, but cancer vaccine trials have not been successful in late-stage patients with treatment-refractory tumours1,2. We are testing melanoma FixVac (BNT111)-an intravenously administered liposomal RNA (RNA-LPX) vaccine, which targets four non-mutated, tumour-associated antigens that are prevalent in melanoma-in an ongoing, first-in-human, dose-escalation phase I trial in patients with advanced melanoma (Lipo-MERIT trial, ClinicalTrials.gov identifier NCT02410733). We report here data from an exploratory interim analysis that show that melanoma FixVac, alone or in combination with blockade of the checkpoint inhibitor PD1, mediates durable objective responses in checkpoint-inhibitor (CPI)-experienced patients with unresectable melanoma. Clinical responses are accompanied by the induction of strong CD4+ and CD8+ T cell immunity against the vaccine antigens. The antigen-specific cytotoxic T-cell responses in some responders reach magnitudes typically reported for adoptive T-cell therapy, and are durable. Our findings indicate that RNA-LPX vaccination is a potent immunotherapy in patients with CPI-experienced melanoma, and suggest the general utility of non-mutant shared tumour antigens as targets for cancer vaccination.


Subject(s)
Antineoplastic Agents/therapeutic use , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Melanoma/immunology , Melanoma/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , RNA, Neoplasm/genetics , T-Lymphocytes/immunology , Antigens, Neoplasm/immunology , Antineoplastic Agents/pharmacology , Cancer Vaccines/administration & dosage , Cancer Vaccines/adverse effects , Combined Modality Therapy , Humans , Melanoma/drug therapy , Melanoma/pathology , Neoplasm Staging , T-Lymphocytes/cytology , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology , Vaccination
5.
Nature ; 547(7662): 222-226, 2017 07 13.
Article in English | MEDLINE | ID: mdl-28678784

ABSTRACT

T cells directed against mutant neo-epitopes drive cancer immunity. However, spontaneous immune recognition of mutations is inefficient. We recently introduced the concept of individualized mutanome vaccines and implemented an RNA-based poly-neo-epitope approach to mobilize immunity against a spectrum of cancer mutations. Here we report the first-in-human application of this concept in melanoma. We set up a process comprising comprehensive identification of individual mutations, computational prediction of neo-epitopes, and design and manufacturing of a vaccine unique for each patient. All patients developed T cell responses against multiple vaccine neo-epitopes at up to high single-digit percentages. Vaccine-induced T cell infiltration and neo-epitope-specific killing of autologous tumour cells were shown in post-vaccination resected metastases from two patients. The cumulative rate of metastatic events was highly significantly reduced after the start of vaccination, resulting in a sustained progression-free survival. Two of the five patients with metastatic disease experienced vaccine-related objective responses. One of these patients had a late relapse owing to outgrowth of ß2-microglobulin-deficient melanoma cells as an acquired resistance mechanism. A third patient developed a complete response to vaccination in combination with PD-1 blockade therapy. Our study demonstrates that individual mutations can be exploited, thereby opening a path to personalized immunotherapy for patients with cancer.


Subject(s)
Cancer Vaccines/genetics , Cancer Vaccines/immunology , Melanoma/immunology , Melanoma/therapy , Mutation/genetics , Precision Medicine/methods , RNA/genetics , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , B7-H1 Antigen/immunology , CD8 Antigens/immunology , Cancer Vaccines/therapeutic use , Epitopes/genetics , Epitopes/immunology , Humans , Immunotherapy/methods , Melanoma/genetics , Neoplasm Metastasis , Neoplasm Recurrence, Local/prevention & control , Nivolumab , Programmed Cell Death 1 Receptor/antagonists & inhibitors , T-Lymphocytes/immunology , Vaccination , beta 2-Microglobulin/deficiency
6.
J Immunother ; 40(5): 187-195, 2017 06.
Article in English | MEDLINE | ID: mdl-28441264

ABSTRACT

B-cell acute lymphoblastic leukemia (B-ALL) is the commonest childhood cancer and the prognosis of children with relapsed or therapy refractory disease remains a challenge. Treatment with chimeric antigen receptor-modified T cells targeting the CD19 antigen (CART-19 therapy) has been presented as a promising approach toward improving the outcome of relapsed or refractory disease. However, 10%-20% of the patients suffer another relapse. Epitope-loss under therapy pressure has been suggested as a mechanism of tumor cells to escape the recognition from CART-19 therapy. In this work, we analyzed the expression of CD19 isoforms in a cohort of 14 children with CD19 B-ALL and 6 nonleukemia donors. We showed that an alternatively spliced CD19 mRNA isoform lacking exon 2, and therefore the CART-19 epitope, but not isoforms lacking the transmembrane and cytosolic domains are expressed in leukemic blasts at diagnosis in children and in the bone marrow of nonleukemia donors. Furthermore, we clarified the sequence of a further isoform lacking the epitope recognized by CART-19 therapy and disclosed the presence of new isoforms. In comparison with the children, we showed that alternatively spliced CD19 mRNA isoforms affecting exon 2 are also expressed in 6 adult patients with CD19 B-ALL. On top of that, one of the adults expressed an isoform lacking the CD19 transmembrane and cytosolic domains. In conclusion, we proved that some of the CD19 isoforms contributing to CART-19 escape already preexist at diagnosis and could evolve as a dominant clone during CART-19 therapy suggesting the application of combined treatment approaches.


Subject(s)
Antigens, CD19/metabolism , Cancer Vaccines/immunology , Epitopes, T-Lymphocyte/metabolism , Immunotherapy, Adoptive/methods , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Protein Isoforms/metabolism , T-Lymphocytes/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Antigens, CD19/genetics , Child , Child, Preschool , Cohort Studies , Epitopes, T-Lymphocyte/genetics , Female , Humans , Infant , Infant, Newborn , Male , Middle Aged , Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Protein Isoforms/genetics , Receptors, Antigen, T-Cell/genetics , Recombinant Fusion Proteins/genetics , T-Cell Antigen Receptor Specificity , T-Lymphocytes/transplantation , Treatment Outcome , Tumor Escape , Young Adult
7.
Nature ; 534(7607): 396-401, 2016 06 16.
Article in English | MEDLINE | ID: mdl-27281205

ABSTRACT

Lymphoid organs, in which antigen presenting cells (APCs) are in close proximity to T cells, are the ideal microenvironment for efficient priming and amplification of T-cell responses. However, the systemic delivery of vaccine antigens into dendritic cells (DCs) is hampered by various technical challenges. Here we show that DCs can be targeted precisely and effectively in vivo using intravenously administered RNA-lipoplexes (RNA-LPX) based on well-known lipid carriers by optimally adjusting net charge, without the need for functionalization of particles with molecular ligands. The LPX protects RNA from extracellular ribonucleases and mediates its efficient uptake and expression of the encoded antigen by DC populations and macrophages in various lymphoid compartments. RNA-LPX triggers interferon-α (IFNα) release by plasmacytoid DCs and macrophages. Consequently, DC maturation in situ and inflammatory immune mechanisms reminiscent of those in the early systemic phase of viral infection are activated. We show that RNA-LPX encoding viral or mutant neo-antigens or endogenous self-antigens induce strong effector and memory T-cell responses, and mediate potent IFNα-dependent rejection of progressive tumours. A phase I dose-escalation trial testing RNA-LPX that encode shared tumour antigens is ongoing. In the first three melanoma patients treated at a low-dose level, IFNα and strong antigen-specific T-cell responses were induced, supporting the identified mode of action and potency. As any polypeptide-based antigen can be encoded as RNA, RNA-LPX represent a universally applicable vaccine class for systemic DC targeting and synchronized induction of both highly potent adaptive as well as type-I-IFN-mediated innate immune mechanisms for cancer immunotherapy.


Subject(s)
Antigens, Neoplasm/immunology , Antigens, Viral/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Immunotherapy/methods , Melanoma/immunology , Melanoma/therapy , RNA/administration & dosage , Administration, Intravenous , Animals , Antigen Presentation/immunology , Antigens, Neoplasm/genetics , Antigens, Viral/genetics , Autoantigens/genetics , Autoantigens/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/genetics , Clinical Trials, Phase I as Topic , Dendritic Cells/cytology , Disease Models, Animal , Drug Carriers/administration & dosage , Female , Humans , Interferon Type I/immunology , Interferon Type I/metabolism , Lymphocyte Activation/immunology , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Macrophages/immunology , Macrophages/metabolism , Male , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Nanoparticles/administration & dosage , RNA/genetics , Static Electricity , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Toll-Like Receptor 7/immunology
8.
J Immunol ; 194(12): 6177-89, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25957167

ABSTRACT

The systematic assessment of the human immune system bears huge potential to guide rational development of novel immunotherapies and clinical decision making. Multiple assays to monitor the quantity, phenotype, and function of Ag-specific T cells are commonly used to unravel patients' immune signatures in various disease settings and during therapeutic interventions. When compared with tests measuring soluble analytes, cellular immune assays have a higher variation, which is a major technical factor limiting their broad adoption in clinical immunology. The key solution may arise from continuous control of assay performance using TCR-engineered reference samples. We developed a simple, stable, robust, and scalable technology to generate reference samples that contain defined numbers of functional Ag-specific T cells. First, we show that RNA-engineered lymphocytes, equipped with selected TCRs, can repetitively deliver functional readouts of a controlled size across multiple assay platforms. We further describe a concept for the application of TCR-engineered reference samples to keep assay performance within or across institutions under tight control. Finally, we provide evidence that these novel control reagents can sensitively detect assay variation resulting from typical sources of error, such as low cell quality, loss of reagent stability, suboptimal hardware settings, or inaccurate gating.


Subject(s)
Immunologic Tests/methods , Immunologic Tests/standards , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Gene Expression , Genetic Engineering , HLA Antigens/chemistry , HLA Antigens/immunology , Humans , Immunotherapy/methods , Peptides/chemistry , Peptides/immunology , Protein Multimerization , T-Cell Antigen Receptor Specificity/genetics , T-Cell Antigen Receptor Specificity/immunology
9.
Cancer Immunol Immunother ; 64(5): 585-98, 2015 May.
Article in English | MEDLINE | ID: mdl-25854580

ABSTRACT

Multiparameter flow cytometry is an indispensable method for assessing antigen-specific T cells in basic research and cancer immunotherapy. Proficiency panels have shown that cell sample processing, test protocols and data analysis may all contribute to the variability of the results obtained by laboratories performing ex vivo T cell immune monitoring. In particular, analysis currently relies on a manual, step-by-step strategy employing serial gating decisions based on visual inspection of one- or two-dimensional plots. It is therefore operator dependent and subjective. In the context of continuing efforts to support inter-laboratory T cell assay harmonization, the CIMT Immunoguiding Program organized its third proficiency panel dedicated to the detection of antigen-specific CD8(+) T cells by HLA-peptide multimer staining. We first assessed the contribution of manual data analysis to the variability of reported T cell frequencies within a group of laboratories staining and analyzing the same cell samples with their own reagents and protocols. The results show that data analysis is a source of variation in the multimer assay outcome. To evaluate whether an automated analysis approach can reduce variability of proficiency panel data, we used a hierarchical statistical mixture model to identify cell clusters. Challenges for automated analysis were the need to process non-standardized data sets from multiple centers, and the fact that the antigen-specific cell frequencies were very low in most samples. We show that this automated method can circumvent difficulties inherent to manual gating strategies and is broadly applicable for experiments performed with heterogeneous protocols and reagents.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Flow Cytometry/statistics & numerical data , HLA Antigens/analysis , Monitoring, Immunologic/methods , CD8-Positive T-Lymphocytes/cytology , Data Interpretation, Statistical , Electronic Data Processing , Flow Cytometry/standards , Healthy Volunteers , Humans , Observer Variation , Reproducibility of Results
10.
Cancer Immunol Res ; 2(12): 1230-44, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25245536

ABSTRACT

The determination of the epitope specificity of disease-associated T-cell responses is relevant for the development of biomarkers and targeted immunotherapies against cancer, autoimmune, and infectious diseases. The lack of known T-cell epitopes and corresponding T-cell receptors (TCR) for novel antigens hinders the efficient development and monitoring of new therapies. We developed an integrated approach for the systematic retrieval and functional characterization of TCRs from single antigen-reactive T cells that includes the identification of epitope specificity. This is accomplished through the rapid cloning of full-length TCR-α and TCR-ß chains directly from single antigen-specific CD8(+) or CD4(+) T lymphocytes. The functional validation of cloned TCRs is conducted using in vitro-transcribed RNA transfer for expression of TCRs in T cells and HLA molecules in antigen-presenting cells. This method avoids the work and bias associated with repetitive cycles of in vitro T-cell stimulation, and enables fast characterization of antigen-specific T-cell responses. We applied this strategy to viral and tumor-associated antigens (TAA), resulting in the retrieval of 56 unique functional antigen-specific TCRs from human CD8(+) and CD4(+) T cells (13 specific for CMV-pp65, 16 specific for the well-known TAA NY-ESO-1, and 27 for the novel TAA TPTE), which are directed against 39 different epitopes. The proof-of-concept studies with TAAs NY-ESO-1 and TPTE revealed multiple novel TCR specificities. Our approach enables the rational development of immunotherapy strategies by providing antigen-specific TCRs and immunogenic epitopes.


Subject(s)
Epitopes, T-Lymphocyte/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Animals , Antigens, Neoplasm/immunology , Cell Line , Cloning, Molecular , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Humans , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/immunology , Phosphoproteins/immunology , Protein Binding , Receptors, Antigen, T-Cell/genetics , Viral Matrix Proteins/immunology
11.
Cancer Immunol Immunother ; 63(12): 1273-84, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25164876

ABSTRACT

Immunotherapeutic approaches are emerging as promising new treatment options for patients with solid cancers. The host immune system in cancer patients is dysfunctional due to a number of reasons. The level of immunosuppression is variable at the time of diagnosis and depends on the particular cancer entity, stage, and prior anti-cancer therapies. For many cancer entities, the immune alterations of the respective patient population have not been further characterized even though a patient's immunophenotype may be prognostic for the course of the disease or predictive for clinical/biological response to immunotherapy. In this study, we used flow cytometry to determine the phenotype of peripheral blood mononuclear cells (PBMCs) from 30 patients with heavily pre-treated, advanced adenocarcinoma of the stomach and gastro-esophageal junction. The frequencies and activation status of relevant immune effector populations were determined in PBMCs and compared to those of healthy individuals. This report provides comprehensive immune phenotyping data of a patient population with a high medical need.


Subject(s)
Adenocarcinoma/blood , Esophagogastric Junction/pathology , Leukocytes, Mononuclear/immunology , Stomach Neoplasms/blood , Adenocarcinoma/immunology , Adult , Aged , Female , Humans , Immunotherapy , Male , Middle Aged , Prognosis , Stomach Neoplasms/immunology
12.
Cytometry A ; 85(7): 621-7, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24616430

ABSTRACT

Staining of transcription factors (TFs) together with retention of fluorescent reporter proteins is hindered by loss of fluorescence using current available methods. In this study, it is shown that current TF staining protocols do not destroy fluorescent proteins (FPs) but rather that fixation is not sufficient to retain FPs in the cytosol of the permeabilized cells. In this article, a simple and reliable protocol is elaborated, which allows efficient TF and cytokine staining while retaining FPs inside fixed cells.


Subject(s)
Cytokines/analysis , Flow Cytometry/methods , Nuclear Proteins/analysis , Transcription Factors/analysis , Animals , Cytoplasm/metabolism , Fixatives , Fluorescent Dyes , Forkhead Transcription Factors , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mycobacterium tuberculosis/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3 , Staining and Labeling , T-Box Domain Proteins , T-Lymphocytes/cytology , Tissue Fixation/methods
13.
Hum Vaccin Immunother ; 9(9): 2025-32, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23877042

ABSTRACT

The 11th Annual Meeting of Association for Cancer Immunotherapy (CIMT) welcomed more than 700 scientists around the world to Mainz, Germany and continued to be the largest immunotherapy meeting in Europe. Renowned speakers from various fields of cancer immunotherapy gave lectures under CIMT2013's tag: "Advancing targeted therapies" the highlights of which are summarized in this meeting report.


Subject(s)
Cancer Vaccines/therapeutic use , Cell Transplantation/methods , Combined Modality Therapy/methods , Immunotherapy/methods , Molecular Targeted Therapy/methods , Neoplasms/therapy , Cell Transplantation/trends , Combined Modality Therapy/trends , Humans , Immunotherapy/trends , Molecular Targeted Therapy/trends , Neoplasms/immunology
14.
Cancer Immunol Immunother ; 62(4): 615-27, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23138872

ABSTRACT

Robust and sensitive ELISPOT protocols are commonly applied concomitant with the development of new immunotherapeutics. Despite the knowledge that individual serum batches differ in their composition and may change properties over time, serum is still commonly used in immunologic assays. Commercially available serum batches are expensive, limited in quantity and need to be pretested for suitability in immunologic assays, which is a laborious process. The aim of this study was to test whether serum-free freezing media can lead to high cell viability and favorable performance across multiple ELISPOT assay protocols. Thirty-one laboratories from ten countries participated in a proficiency panel organized by the Cancer Immunotherapy Immunoguiding Program to test the influence of different freezing media on cell quality and immunologic function. Each center received peripheral blood mononuclear cells which were frozen in three different media. The participants were asked to quantify antigen-specific CD8+ T-cell responses against model antigens using their locally established IFN-gamma ELISPOT protocols. Self-made and commercially available serum-free freezing media led to higher cell viability and similar cell recovery after thawing and resting compared to freezing media supplemented with human serum. Furthermore, the test performance as determined by (1) background spot production, (2) replicate variation, (3) frequency of detected antigen-specific spots and (4) response detection rate was similar for serum and serum-free conditions. We conclude that defined and accessible serum-free freezing media should be recommended for freezing cells stored for subsequent ELISPOT analysis.


Subject(s)
Culture Media, Serum-Free/chemistry , Enzyme-Linked Immunospot Assay/methods , Leukocytes, Mononuclear/chemistry , Animals , Cattle , Cell Survival , Enzyme-Linked Immunospot Assay/standards , Epitopes, T-Lymphocyte/analysis , Epitopes, T-Lymphocyte/immunology , Freezing , HLA-A2 Antigen/chemistry , HLA-A2 Antigen/immunology , Humans , Interferon-gamma/chemistry , Interferon-gamma/immunology , Leukocytes, Mononuclear/immunology , Peptide Fragments/chemistry , Peptide Fragments/immunology , T-Lymphocytes/chemistry , T-Lymphocytes/immunology
15.
Cancer Immunol Res ; 1(6): 386-92, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24778131

ABSTRACT

Vaccination with in vitro transcribed RNA encoding tumor antigens is an emerging approach in cancer immunotherapy. Attempting to further improve RNA vaccine efficacy, we have explored combining RNA with immunomodulators such as rapamycin. Rapamycin, the inhibitor of mTOR, was used originally for immunosuppression. Recent reports in mouse systems, however, suggest that mTOR inhibition may enhance the formation and differentiation of the memory CD8(+) T-cell pool. Because memory T-cell formation is critical to the outcome of vaccination approaches, we studied the impact of rapamycin on the in vivo primed RNA vaccine-induced immune response using the chicken ovalbumin-expressing B16 melanoma model in C57BL/6 mice. Our data show that treatment with rapamycin at the effector-to-memory transition phase skews the vaccine-induced immune response toward the formation of a quantitatively and qualitatively superior memory pool and results in a better recall response. Tumor-infiltrating immune cells from these mice display a favorable ratio of effector versus suppressor cell populations. Survival of mice treated with the combined regimen of RNA vaccination with rapamycin is significantly longer (91.5 days) than that in the control groups receiving only one of these compounds (32 and 46 days, respectively). Our findings indicate that rapamycin enhances therapeutic efficacy of antigen-specific CD8(+) T cells induced by RNA vaccination, and we propose further clinical exploration of rapamycin as a component of immunotherapeutic regimens.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Cancer Vaccines/therapeutic use , Melanoma, Experimental/drug therapy , Sirolimus/therapeutic use , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Cell Differentiation/drug effects , Cell Differentiation/immunology , Combined Modality Therapy , Drug Screening Assays, Antitumor/methods , Female , Immunologic Memory , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma, Experimental/immunology , Mice, Inbred C57BL , RNA, Neoplasm/immunology , Vaccination/methods , Vaccines, Synthetic/immunology , Vaccines, Synthetic/therapeutic use
16.
Int J Cancer ; 131(1): 140-9, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-21858810

ABSTRACT

CD4(+) T cells have been shown to be crucial for the induction and maintenance of cytotoxic T cell responses and to be also capable of mediating direct tumor rejection. Therefore, the anticancer therapeutic efficacy of peptide-based vaccines may be improved by addition of HLA class II epitopes to stimulate T helper cells. Survivin is an apoptosis inhibiting protein frequently overexpressed in tumors. Here we describe the first immunological evaluation of a survivin-derived CD4(+) T cell epitope in a multipeptide immunotherapy trial for prostate carcinoma patients. The survivin peptide is promiscuously presented by several human HLA-DRB1 molecules and, most importantly, is naturally processed by dendritic cells. In vaccinated patients, it was able to induce frequent, robust and multifunctional CD4(+) T cell responses, as monitored by IFN-γ ELISPOT and intracellular cytokine staining. Thus, this HLA-DR restricted epitope is broadly immunogenic and should be valuable for stimulating T helper cells in patients suffering from a wide range of tumors.


Subject(s)
Cancer Vaccines/immunology , Inhibitor of Apoptosis Proteins/immunology , Lymphocyte Activation , Prostatic Neoplasms/immunology , Prostatic Neoplasms/therapy , T-Lymphocytes, Helper-Inducer/immunology , Aged , Antigens, Neoplasm/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/therapeutic use , Cytokines/biosynthesis , Dendritic Cells/immunology , Dendritic Cells/metabolism , Epitopes, T-Lymphocyte/immunology , HLA-DRB1 Chains/immunology , Humans , Interferon-gamma/biosynthesis , Male , Middle Aged , Peptide Fragments/immunology , Survivin , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Helper-Inducer/metabolism
17.
Cancer Res ; 71(19): 6132-42, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21816907

ABSTRACT

Intranodal immunization with antigen-encoding naked RNA may offer a simple and safe approach to induce antitumor immunity. RNA taken up by nodal dendritic cells (DC) coactivates toll-like receptor (TLR) signaling that will prime and expand antigen-specific T cells. In this study, we show that RNA vaccination can be optimized by coadministration of the DC-activating Fms-like tyrosine kinase 3 (FLT3) ligand as an effective adjuvant. Systemic administration of FLT3 ligand prior to immunization enhanced priming and expansion of antigen-specific CD8(+) T cells in lymphoid organs, T-cell homing into melanoma tumors, and therapeutic activity of the intranodal RNA. Unexpectedly, plasmacytoid DCs (pDC) were found to be essential for the adjuvant effect of FLT3 ligand and they were systemically expanded together with conventional DCs after treatment. In response to FLT3 ligand, pDCs maintained an immature phenotype, internalized RNA, and presented the RNA-encoded antigen for efficient induction of antigen-specific CD8(+) T-cell responses. Coadministration of FLT3 ligand with RNA vaccination achieved remarkable cure rates and survival of mice with advanced melanoma. Our findings show how to improve the simple and safe strategy offered by RNA vaccines for cancer immunotherapy.


Subject(s)
Cancer Vaccines , Melanoma/therapy , Membrane Proteins/immunology , RNA/immunology , Adjuvants, Immunologic/administration & dosage , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Humans , Immunotherapy/methods , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Melanoma/immunology , Membrane Proteins/administration & dosage , Mice , Neoplasm Transplantation , RNA/administration & dosage
18.
J Transl Med ; 9: 108, 2011 Jul 11.
Article in English | MEDLINE | ID: mdl-21745365

ABSTRACT

BACKGROUND: The introduction of antibody markers to identify undesired cell populations in flow-cytometry based assays, so called DUMP channel markers, has become a practice in an increasing number of labs performing HLA-peptide multimer assays. However, the impact of the introduction of a DUMP channel in multimer assays has so far not been systematically investigated across a broad variety of protocols. METHODS: The Cancer Research Institute's Cancer Immunotherapy Consortium (CRI-CIC) conducted a multimer proficiency panel with a specific focus on the impact of DUMP channel use. The panel design allowed individual laboratories to use their own protocol for thawing, staining, gating, and data analysis. Each experiment was performed twice and in parallel, with and without the application of a dump channel strategy. RESULTS: The introduction of a DUMP channel is an effective measure to reduce the amount of non-specific MULTIMER binding to T cells. Beneficial effects for the use of a DUMP channel were observed across a wide range of individual laboratories and for all tested donor-antigen combinations. In 48% of experiments we observed a reduction of the background MULTIMER-binding. In this subgroup of experiments the median background reduction observed after introduction of a DUMP channel was 0.053%. CONCLUSIONS: We conclude that appropriate use of a DUMP channel can significantly reduce background staining across a large fraction of protocols and improve the ability to accurately detect and quantify the frequency of antigen-specific T cells by multimer reagents. Thus, use of a DUMP channel may become crucial for detecting low frequency antigen-specific immune responses. Further recommendations on assay performance and data presentation guidelines for publication of MULTIMER experimental data are provided.


Subject(s)
Biomarkers/metabolism , HLA Antigens/metabolism , Peptides/metabolism , Protein Multimerization , Staining and Labeling/methods , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Survival , Coloring Agents/metabolism , Computational Biology , Humans , Lymphocyte Count , Protein Binding , Reproducibility of Results , Subcellular Fractions/metabolism
19.
Cancer Res ; 69(21): 8412-9, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19843860

ABSTRACT

Renal cell carcinoma is frequently infiltrated by cells of the immune system. This makes it important to understand interactions between cancer cells and immune cells so they can be manipulated to bring clinical benefit. Here, we analyze subsets and functions of T lymphocytes infiltrating renal cell tumors directly ex vivo following mechanical disaggregation and without any culture step. Subpopulations of memory and effector CD4(+) Th1, Th2, and Th17 and CD8(+) Tc1 cells were identified based on surface phenotype, activation potential, and multicytokine production. Compared with the same patient's peripheral blood, T lymphocytes present inside tumors were found to be enriched in functional CD4(+) cells of the Th1 lineage and in effector memory CD8(+) cells. Additionally, several populations of CD4(+) and CD8(+) regulatory T cells were identified that may synergize to locally dampen antitumor T-cell responses.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Renal Cell/immunology , Kidney Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , T-Lymphocytes, Regulatory/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Female , Humans , Immunologic Memory , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Male , Middle Aged , Phenotype , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology , Th1 Cells/immunology , Th1 Cells/metabolism , Th1 Cells/pathology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...